1
|
Andraski AB, Sacks FM, Aikawa M, Singh SA. Understanding HDL Metabolism and Biology Through In Vivo Tracer Kinetics. Arterioscler Thromb Vasc Biol 2024; 44:76-88. [PMID: 38031838 PMCID: PMC10842918 DOI: 10.1161/atvbaha.123.319742] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023]
Abstract
HDL (high-density lipoprotein), owing to its high protein content and small size, is the densest circulating lipoprotein. In contrast to lipid-laden VLDL (very-low-density lipoprotein) and LDL (low-density lipoprotein) that promote atherosclerosis, HDL is hypothesized to mitigate atherosclerosis via reverse cholesterol transport, a process that entails the uptake and clearance of excess cholesterol from peripheral tissues. This process is mediated by APOA1 (apolipoprotein A-I), the primary structural protein of HDL, as well as by the activities of additional HDL proteins. Tracer-dependent kinetic studies are an invaluable tool to study HDL-mediated reverse cholesterol transport and overall HDL metabolism in humans when a cardiovascular disease therapy is investigated. Unfortunately, HDL cholesterol-raising therapies have not been successful at reducing cardiovascular events suggesting an incomplete picture of HDL biology. However, as HDL tracer studies have evolved from radioactive isotope- to stable isotope-based strategies that in turn are reliant on mass spectrometry technologies, the complexity of the HDL proteome and its metabolism can be more readily addressed. In this review, we outline the motivations, timelines, advantages, and disadvantages of the various tracer kinetics strategies. We also feature the metabolic properties of select HDL proteins known to regulate reverse cholesterol transport, which in turn underscore that HDL lipoproteins comprise a heterogeneous particle population whose distinct protein constituents and kinetics likely determine its function and potential contribution to cholesterol clearance.
Collapse
Affiliation(s)
- Allison B. Andraski
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Frank M. Sacks
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Sasha A. Singh
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
2
|
Fu Q, Murray CI, Karpov OA, Van Eyk JE. Automated proteomic sample preparation: The key component for high throughput and quantitative mass spectrometry analysis. MASS SPECTROMETRY REVIEWS 2023; 42:873-886. [PMID: 34786750 PMCID: PMC10339360 DOI: 10.1002/mas.21750] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 10/11/2021] [Accepted: 10/27/2021] [Indexed: 06/13/2023]
Abstract
Sample preparation for mass spectrometry-based proteomics has many tedious and time-consuming steps that can introduce analytical errors. In particular, the steps around the proteolytic digestion of protein samples are prone to inconsistency. One route for reliable sample processing is the development and optimization of a workflow utilizing an automated liquid handling workstation. Diligent assessment of the sample type, protocol design, reagents, and incubation conditions can significantly improve the speed and consistency of preparation. When combining robust liquid chromatography-mass spectrometry with either discovery or targeted methods, automated sample preparation facilitates increased throughput and reproducible quantitation of biomarker candidates. These improvements in analysis are also essential to process the large patient cohorts necessary to validate a candidate biomarker for potential clinical use. This article reviews the steps in the workflow, optimization strategies, and known applications in clinical, pharmaceutical, and research fields that demonstrate the broad utility for improved automation of sample preparation in the proteomic field.
Collapse
Affiliation(s)
- Qin Fu
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Christopher I Murray
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Oleg A Karpov
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jennifer E Van Eyk
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
3
|
Ortiz-Riaño EJ, Mancera-Zapata DL, Ulloa-Ramírez M, Arce-Vega F, Morales-Narváez E. Measurement of Protein Kinetics Using a Liquid Phase-Based Biosensing Platform. Anal Chem 2022; 94:15553-15557. [PMID: 36253365 DOI: 10.1021/acs.analchem.2c03305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Macromolecular association is crucial to many fields in biomedical sciences, including drug development, gene editing, and diagnostics. In particular, protein-protein association and dissociation rate constants are typically determined using surface plasmon resonance systems, which require costly instrumentation and cumbersome procedures (e.g., blocking, washing, and separation). Herein, we demonstrate that protein-binding constants can be readily determined using a real-time biosensing platform facilitated by graphene oxide-modified microwell plates and fluorophore-labeled proteins, where the fluorescent probes remain highly fluorescent during protein association, whereas fluorescent bioprobes that are not associated with their counterparts are quenched by graphene oxide. Binding data of three pairs of proteins were systematically determined employing this single-step platform and compared with those data reported by the suppliers or the literature, suggesting that this approach is comparable and consistent with the existing ones. Such pairs include (i) human immunoglobulin G (H-IgG)-fluorophore-labeled anti-H-IgG, (ii) prostate-specific antigen (PSA)-quantum dot-labeled anti-PSA, and (iii) anti-RBD-fluorophore-labeled SARS-CoV-2 spike receptor-binding domain recombinant protein. We also offer an open-source software that automatically determines the binding kinetics constants of proteins. This Technical Note introduces a simple, yet effective, platform to determine relevant information on protein kinetics, which can be performed using a microwell plate reader and economical materials like graphene oxide. We foresee a new generation of diagnostics based on our affordable protein kinetics analysis.
Collapse
Affiliation(s)
- Edwin J Ortiz-Riaño
- Biophotonic Nanosensors Laboratory, Centro de Investigaciones en Óptica, A. C., Loma del Bosque 115, Lomas del Campestre, León, Guanajuato37150, Mexico
| | - Diana L Mancera-Zapata
- Biophotonic Nanosensors Laboratory, Centro de Investigaciones en Óptica, A. C., Loma del Bosque 115, Lomas del Campestre, León, Guanajuato37150, Mexico
| | - Martha Ulloa-Ramírez
- Biophotonic Nanosensors Laboratory, Centro de Investigaciones en Óptica, A. C., Loma del Bosque 115, Lomas del Campestre, León, Guanajuato37150, Mexico.,Universidad de Guadalajara, Guadalajara44100, Jalisco, Mexico
| | - Fernando Arce-Vega
- Centro de Investigaciones en Óptica, A. C., Loma del Bosque 115, Lomas del Campestre, León37150, Guanajuato, Mexico
| | - Eden Morales-Narváez
- Biophotonic Nanosensors Laboratory, Centro de Investigaciones en Óptica, A. C., Loma del Bosque 115, Lomas del Campestre, León, Guanajuato37150, Mexico
| |
Collapse
|
4
|
Dandan M, Han J, Mann S, Kim R, Mohammed H, Nyangau E, Hellerstein M. Turnover Rates of the Low-Density Lipoprotein Receptor and PCSK9: Added Dimension to the Cholesterol Homeostasis Model. Arterioscler Thromb Vasc Biol 2021; 41:2866-2876. [PMID: 34615375 DOI: 10.1161/atvbaha.121.316764] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We measured the turnover rates of the LDLR (low-density lipoprotein receptor) and PCSK9 (proprotein convertase subtilisin/kexin type 9) in mice by metabolic labeling with heavy water and mass spectrometry. Approach and Results: In liver of mice fed high-cholesterol diets, LDLR mRNA levels and synthesis rates were markedly lower with complete suppression of cholesterol synthesis and higher cholesterol content, consistent with the Brown-Goldstein model of tissue cholesterol homeostasis. We observed markedly lower PCSK9 mRNA levels and synthesis rates in liver and lower concentrations and synthesis rates in plasma. Hepatic LDLR half-life (t½) was prolonged, consistent with an effect of reduced PCSK9, and resulted in no reduction in hepatic LDLR content despite reduced mRNA levels and LDLR synthesis rates. These changes in PCSK9 synthesis complement and expand the well-established model of tissue cholesterol homeostasis in mouse liver, in that reduced synthesis and levels of PCSK9 counterbalance lower LDLR synthesis by promoting less LDLR catabolism, thereby maintaining uptake of LDL cholesterol into liver despite high intracellular cholesterol concentrations. CONCLUSIONS Lower hepatic synthesis and secretion of PCSK9, an SREBP2 (sterol response element binding protein) target gene, results in longer hepatic LDLR t½ in response to cholesterol feeding in mice in the face of high intracellular cholesterol content. PCSK9 modulation opposes the canonical lowering of LDLR mRNA and synthesis by cholesterol surplus and preserves LDLR levels. The physiological and therapeutic implications of these opposing control mechanisms over liver LDLR are of interest and may reflect subservience of hepatic cholesterol homeostasis to whole body cholesterol needs.
Collapse
Affiliation(s)
- Mohamad Dandan
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley
| | - Julia Han
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley
| | - Sabrina Mann
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley
| | - Rachael Kim
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley
| | - Hussein Mohammed
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley
| | - Edna Nyangau
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley
| | - Marc Hellerstein
- Graduate Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley
| |
Collapse
|
5
|
Oleaga C, Shapiro MD, Hay J, Mueller PA, Miles J, Huang C, Friz E, Tavori H, Toth PP, Wójcik C, Warden BA, Purnell JQ, Duell PB, Pamir N, Fazio S. Hepatic Sensing Loop Regulates PCSK9 Secretion in Response to Inhibitory Antibodies. J Am Coll Cardiol 2021; 78:1437-1449. [PMID: 34593126 PMCID: PMC8486917 DOI: 10.1016/j.jacc.2021.07.056] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/08/2021] [Accepted: 07/26/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND Monoclonal antibodies against proprotein convertase subtilisin/kexin type 9 (PCSK9i) lower LDL-C by up to 60% and increase plasma proprotein convertase subtilisin/kexin type 9 (PCSK9) levels by 10-fold. OBJECTIVES The authors studied the reasons behind the robust increase in plasma PCSK9 levels by testing the hypothesis that mechanisms beyond clearance via the low-density lipoprotein receptor (LDLR) contribute to the regulation of cholesterol homeostasis. METHODS In clinical cohorts, animal models, and cell-based studies, we measured kinetic changes in PCSK9 production and clearance in response to PCSK9i. RESULTS In a patient cohort receiving PCSK9i therapy, plasma PCSK9 levels rose 11-fold during the first 3 months and then plateaued for 15 months. In a cohort of healthy volunteers, a single injection of PCSK9i increased plasma PCSK9 levels within 12 hours; the rise continued for 9 days until it plateaued at 10-fold above baseline. We recapitulated the rapid rise in PCSK9 levels in a mouse model, but only in the presence of LDLR. In vivo turnover and in vitro pulse-chase studies identified 2 mechanisms contributing to the rapid increase in plasma PCSK9 levels in response to PCSK9i: 1) the expected delayed clearance of the antibody-bound PCSK9; and 2) the unexpected post-translational increase in PCSK9 secretion. CONCLUSIONS PCSK9 re-entry to the liver via LDLR triggers a sensing loop regulating PCSK9 secretion. PCSK9i therapy enhances the secretion of PCSK9, an effect that contributes to the increased plasma PCSK9 levels in treated subjects.
Collapse
Affiliation(s)
- Carlota Oleaga
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Michael D Shapiro
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Joshua Hay
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Paul A Mueller
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Joshua Miles
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Cecilia Huang
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Emily Friz
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Hagai Tavori
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Peter P Toth
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University, and School of Medicine, Baltimore, Maryland, USA; CGH Medical Center, Sterling, Illinois, USA
| | - Cezary Wójcik
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Bruce A Warden
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Jonathan Q Purnell
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - P Barton Duell
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| | - Nathalie Pamir
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA.
| | - Sergio Fazio
- Knight Cardiovascular Institute, Center for Preventive Cardiology, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
6
|
Kapitanov GI, Chabot JR, Narula J, Roy M, Neubert H, Palandra J, Farrokhi V, Johnson JS, Webster R, Jones HM. A Mechanistic Site-Of-Action Model: A Tool for Informing Right Target, Right Compound, And Right Dose for Therapeutic Antagonistic Antibody Programs. FRONTIERS IN BIOINFORMATICS 2021; 1:731340. [DOI: 10.3389/fbinf.2021.731340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
Quantitative modeling is increasingly utilized in the drug discovery and development process, from the initial stages of target selection, through clinical studies. The modeling can provide guidance on three major questions–is this the right target, what are the right compound properties, and what is the right dose for moving the best possible candidate forward. In this manuscript, we present a site-of-action modeling framework which we apply to monoclonal antibodies against soluble targets. We give a comprehensive overview of how we construct the model and how we parametrize it and include several examples of how to apply this framework for answering the questions postulated above. The utilities and limitations of this approach are discussed.
Collapse
|
7
|
Singh SA, Andraski AB, Higashi H, Lee LH, Ramsaroop A, Sacks FM, Aikawa M. Metabolism of PLTP, CETP, and LCAT on multiple HDL sizes using the Orbitrap Fusion Lumos. JCI Insight 2021; 6:143526. [PMID: 33351780 PMCID: PMC7934878 DOI: 10.1172/jci.insight.143526] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 12/16/2020] [Indexed: 11/18/2022] Open
Abstract
Recent in vivo tracer studies demonstrated that targeted mass spectrometry (MS) on the Q Exactive Orbitrap could determine the metabolism of HDL proteins 100s-fold less abundant than apolipoprotein A1 (APOA1). In this study, we demonstrate that the Orbitrap Lumos can measure tracer in proteins whose abundances are 1000s-fold less than APOA1, specifically the lipid transfer proteins phospholipid transfer protein (PLTP), cholesterol ester transfer protein (CETP), and lecithin-cholesterol acyl transferase (LCAT). Relative to the Q Exactive, the Lumos improved tracer detection by reducing tracer enrichment compression, thereby providing consistent enrichment data across multiple HDL sizes from 6 participants. We determined by compartmental modeling that PLTP is secreted in medium and large HDL (alpha2, alpha1, and alpha0) and is transferred from medium to larger sizes during circulation from where it is catabolized. CETP is secreted mainly in alpha1 and alpha2 and remains in these sizes during circulation. LCAT is secreted mainly in medium and small HDL (alpha2, alpha3, prebeta). Unlike PLTP and CETP, LCAT’s appearance on HDL is markedly delayed, indicating that LCAT may reside for a time outside of systemic circulation before attaching to HDL in plasma. The determination of these lipid transfer proteins’ unique metabolic structures was possible due to advances in MS technologies.
Collapse
Affiliation(s)
- Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Allison B Andraski
- Department of Nutrition and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Hideyuki Higashi
- Center for Interdisciplinary Cardiovascular Sciences, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lang Ho Lee
- Center for Interdisciplinary Cardiovascular Sciences, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ashisha Ramsaroop
- Center for Interdisciplinary Cardiovascular Sciences, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Frank M Sacks
- Department of Nutrition and Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Channing Division of Network Medicine, Department of Medicine, and
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Channing Division of Network Medicine, Department of Medicine, and.,Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
8
|
Lassman ME, Chappell DL, McAvoy T, Cheng A, de Alwis DP, Pruitt SK, Laterza OF, Li C, Stoch A, Mayawala K. Experimental Medicine Study to Measure Immune Checkpoint Receptors PD-1 and GITR Turnover Rates In Vivo in Humans. Clin Pharmacol Ther 2020; 109:1575-1582. [PMID: 33280089 DOI: 10.1002/cpt.2129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/12/2020] [Indexed: 11/12/2022]
Abstract
Development of monoclonal antibodies (mAbs) targeting immune-checkpoint receptors (IMRs) for the treatment of cancer is one of the most active areas of investment in the biopharmaceutical industry. A key decision in the clinical development of anti-IMR mAbs is dose selection. Dose selection can be challenging because the traditional oncology paradigm of administering the maximum tolerated dose is not applicable to anti-IMR mAbs. Instead, dose selection should be informed by the pharmacology of immune signaling. Engaging an IMR is a key initial step to triggering pharmacologic effects, and turnover (i.e., the rate of protein synthesis) of the IMR is a key property to determining the dose level needed to engage the IMR. Here, we applied the stable isotope labeling mass spectrometry technique using 13 C6 -leucine to measure the in vivo turnover rates of IMRs in humans. The 13 C6 -leucine was administered to 10 study participants over 15 hours to measure 13 C6 -leucine enrichment kinetics in 2 IMR targets that have been clinically pursued in oncology: GITR and PD-1. We report the first measurements of GITR and PD-1 median half-lives associated with turnover to be 55.6 and ≥ 49.5 hours, respectively. The approach outlined here can be applied to other IMRs and, more generally, to protein targets.
Collapse
Affiliation(s)
- Michael E Lassman
- Translational Molecular Biomarkers, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Derek L Chappell
- Translational Molecular Biomarkers, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Thomas McAvoy
- Translational Molecular Biomarkers, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Amy Cheng
- Translational Medicine Clinical Operations, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Dinesh P de Alwis
- Quantitative Pharmacology and Pharmacometrics, PPDM, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Scott K Pruitt
- Oncology Clinical Research, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Omar F Laterza
- Translational Molecular Biomarkers, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Claire Li
- Quantitative Pharmacology and Pharmacometrics, PPDM, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | | | - Kapil Mayawala
- Quantitative Pharmacology and Pharmacometrics, PPDM, Merck & Co., Inc., Kenilworth, New Jersey, USA
| |
Collapse
|
9
|
Neubert H, Shuford CM, Olah TV, Garofolo F, Schultz GA, Jones BR, Amaravadi L, Laterza OF, Xu K, Ackermann BL. Protein Biomarker Quantification by Immunoaffinity Liquid Chromatography–Tandem Mass Spectrometry: Current State and Future Vision. Clin Chem 2020; 66:282-301. [DOI: 10.1093/clinchem/hvz022] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/12/2019] [Indexed: 12/19/2022]
Abstract
Abstract
Immunoaffinity–mass spectrometry (IA-MS) is an emerging analytical genre with several advantages for profiling and determination of protein biomarkers. Because IA-MS combines affinity capture, analogous to ligand binding assays (LBAs), with mass spectrometry (MS) detection, this platform is often described using the term hybrid methods. The purpose of this report is to provide an overview of the principles of IA-MS and to demonstrate, through application, the unique power and potential of this technology. By combining target immunoaffinity enrichment with the use of stable isotope-labeled internal standards and MS detection, IA-MS achieves high sensitivity while providing unparalleled specificity for the quantification of protein biomarkers in fluids and tissues. In recent years, significant uptake of IA-MS has occurred in the pharmaceutical industry, particularly in the early stages of clinical development, enabling biomarker measurement previously considered unattainable. By comparison, IA-MS adoption by CLIA laboratories has occurred more slowly. Current barriers to IA-MS use and opportunities for expanded adoption are discussed. The path forward involves identifying applications for which IA-MS is the best option compared with LBA or MS technologies alone. IA-MS will continue to benefit from advances in reagent generation, more sensitive and higher throughput MS technologies, and continued growth in use by the broader analytical community. Collectively, the pursuit of these opportunities will secure expanded long-term use of IA-MS for clinical applications.
Collapse
|
10
|
Reyes-Soffer G, Sztalryd C, Horenstein RB, Holleran S, Matveyenko A, Thomas T, Nandakumar R, Ngai C, Karmally W, Ginsberg HN, Ramakrishnan R, Pollin TI. Effects of APOC3 Heterozygous Deficiency on Plasma Lipid and Lipoprotein Metabolism. Arterioscler Thromb Vasc Biol 2019; 39:63-72. [PMID: 30580564 DOI: 10.1161/atvbaha.118.311476] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Objective- Apo (apolipoprotein) CIII inhibits lipoprotein lipase (LpL)-mediated lipolysis of VLDL (very-low-density lipoprotein) triglyceride (TG) and decreases hepatic uptake of VLDL remnants. The discovery that 5% of Lancaster Old Order Amish are heterozygous for the APOC3 R19X null mutation provided the opportunity to determine the effects of a naturally occurring reduction in apo CIII levels on the metabolism of atherogenic containing lipoproteins. Approach and Results- We conducted stable isotope studies of VLDL-TG and apoB100 in 5 individuals heterozygous for the null mutation APOC3 R19X (CT) and their unaffected (CC) siblings. Fractional clearance rates and production rates of VLDL-TG and apoB100 in VLDL, IDL (intermediate-density lipoprotein), LDL, apo CIII, and apo CII were determined. Affected (CT) individuals had 49% reduction in plasma apo CIII levels compared with CCs ( P<0.01) and reduced plasma levels of TG (35%, P<0.02), VLDL-TG (45%, P<0.02), and VLDL-apoB100 (36%, P<0.05). These changes were because of higher fractional clearance rates of VLDL-TG and VLDL-apoB100 with no differences in production rates. CTs had higher rates of the conversion of VLDL remnants to LDL compared with CCs. In contrast, rates of direct removal of VLDL remnants did not differ between the groups. As a result, the flux of apoB100 from VLDL to LDL was not reduced, and the plasma levels of LDL-cholesterol and LDL-apoB100 were not lower in the CT group. Apo CIII production rate was lower in CTs compared with CCs, whereas apo CII production rate was not different between the 2 groups. The fractional clearance rates of both apo CIII and apo CII were higher in CTs than CCs. Conclusions- These studies demonstrate that 50% reductions in plasma apo CIII, in otherwise healthy subjects, results in a significantly higher rate of conversion of VLDL to LDL, with little effect on direct hepatic uptake of VLDL. When put in the context of studies demonstrating significant protection from cardiovascular events in individuals with loss of function variants in the APOC3 gene, our results provide strong evidence that therapies which increase the efficiency of conversion of VLDL to LDL, thereby reducing remnant concentrations, should reduce the risk of cardiovascular disease.
Collapse
Affiliation(s)
- Gissette Reyes-Soffer
- From the Columbia University Vagelos College of Physicians and Surgeons, New York (G.R.-S., S.H., A.M., T.T., R.N., C.N., W.K., H.N.G., R.R.)
| | - Carol Sztalryd
- Maryland School of Medicine, University of Maryland, Baltimore (C.S., R.B.H., T.I.P.)
- Baltimore VA Medical Center, VA Research Service, Geriatric Research, Education and Clinical Center and VA Maryland Health Care System (C.S., T.I.P.)
| | - Richard B Horenstein
- Maryland School of Medicine, University of Maryland, Baltimore (C.S., R.B.H., T.I.P.)
| | - Stephen Holleran
- From the Columbia University Vagelos College of Physicians and Surgeons, New York (G.R.-S., S.H., A.M., T.T., R.N., C.N., W.K., H.N.G., R.R.)
| | - Anastasiya Matveyenko
- From the Columbia University Vagelos College of Physicians and Surgeons, New York (G.R.-S., S.H., A.M., T.T., R.N., C.N., W.K., H.N.G., R.R.)
| | - Tiffany Thomas
- From the Columbia University Vagelos College of Physicians and Surgeons, New York (G.R.-S., S.H., A.M., T.T., R.N., C.N., W.K., H.N.G., R.R.)
| | - Renu Nandakumar
- From the Columbia University Vagelos College of Physicians and Surgeons, New York (G.R.-S., S.H., A.M., T.T., R.N., C.N., W.K., H.N.G., R.R.)
| | - Colleen Ngai
- From the Columbia University Vagelos College of Physicians and Surgeons, New York (G.R.-S., S.H., A.M., T.T., R.N., C.N., W.K., H.N.G., R.R.)
| | - Wahida Karmally
- From the Columbia University Vagelos College of Physicians and Surgeons, New York (G.R.-S., S.H., A.M., T.T., R.N., C.N., W.K., H.N.G., R.R.)
| | - Henry N Ginsberg
- From the Columbia University Vagelos College of Physicians and Surgeons, New York (G.R.-S., S.H., A.M., T.T., R.N., C.N., W.K., H.N.G., R.R.)
| | - Rajasekhar Ramakrishnan
- From the Columbia University Vagelos College of Physicians and Surgeons, New York (G.R.-S., S.H., A.M., T.T., R.N., C.N., W.K., H.N.G., R.R.)
| | - Toni I Pollin
- Maryland School of Medicine, University of Maryland, Baltimore (C.S., R.B.H., T.I.P.)
- Baltimore VA Medical Center, VA Research Service, Geriatric Research, Education and Clinical Center and VA Maryland Health Care System (C.S., T.I.P.)
| |
Collapse
|
11
|
Fan YY, Farrokhi V, Caiazzo T, Wang M, O'Hara DM, Neubert H. Human FcRn Tissue Expression Profile and Half-Life in PBMCs. Biomolecules 2019; 9:biom9080373. [PMID: 31443181 PMCID: PMC6722552 DOI: 10.3390/biom9080373] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 08/08/2019] [Accepted: 08/14/2019] [Indexed: 12/25/2022] Open
Abstract
System-wide quantitative characterization of human neonatal Fc receptor (FcRn) properties is critical for understanding and predicting human PK (pharmacokinetics) as well as the distribution of mAbs and Fc-fusion proteins using PBPK (physiologically-based pharmacokinetic) modeling. To this end, tissue-specific FcRn expression and half-life are important model inputs. Herein, human FcRn tissue expression was measured by peptide immunoaffinity chromatography coupled with high-resolution mass spectrometry. FcRn concentrations across 14 human tissues ranged from low to 230 pmol per gram of tissue. Furthermore, the FcRn half-life was determined to be 11.1 h from a human stable isotope labelled leucine pulse labeling experiment. The spatial and temporal quantitative human FcRn data now promise to enable a refined PBPK model with improved accuracy of human PK predictions for Fc-containing biotherapeutics.
Collapse
Affiliation(s)
- Yao-Yun Fan
- Biomedicine Design, Pfizer Worldwide Research & Development, Andover, MA 01810, USA
| | - Vahid Farrokhi
- Biomedicine Design, Pfizer Worldwide Research & Development, Andover, MA 01810, USA
| | - Teresa Caiazzo
- Biomedicine Design, Pfizer Worldwide Research & Development, Andover, MA 01810, USA
| | - Mengmeng Wang
- Biomedicine Design, Pfizer Worldwide Research & Development, Andover, MA 01810, USA
| | - Denise M O'Hara
- Biomedicine Design, Pfizer Worldwide Research & Development, Andover, MA 01810, USA
| | - Hendrik Neubert
- Biomedicine Design, Pfizer Worldwide Research & Development, Andover, MA 01810, USA.
| |
Collapse
|
12
|
Optimizing hybrid LC-MS/MS binding conditions is critical: impact of biotransformation on quantification of trastuzumab. Bioanalysis 2018; 10:1819-1831. [PMID: 30325201 DOI: 10.4155/bio-2018-0196] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background: Hybrid ligand-binding (LB) LC-MS/MS protein quantitative assays involve a LB step for analyte enrichment that has less stringent requirements than the conventional LB assays. Results: Herceptin™(trastuzumab) binding to HER2 extracellular domain was evaluated using on-bead and off-bead capture formats. The two formats yielded significantly different trastuzumab concentrations in human and monkey serum pharmacokinetic samples. Biotransformations, including deamidation of asparagine and isomerization of aspartic acid near the complementarity-determining regions of trastuzumab, had a profound impact on the LB step for analyte enrichment and trastuzumab quantification. Conclusion: Quantitative measurements were profoundly impacted by LB conditions in a hybrid LB LC-MS/MS protein assay due to biotransformations. Therefore, similar to conventional LB assays, binding conditions should be carefully evaluated during assay development.
Collapse
|
13
|
Perspectives on potentiating immunocapture-LC-MS for the bioanalysis of biotherapeutics and biomarkers. Bioanalysis 2018; 10:1679-1690. [PMID: 30371100 DOI: 10.4155/bio-2018-0205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The integration of ligand-binding assay and LC-MS/MS (immunocapture-LC-MS) has unleashed the combined advantages of both powerful techniques for addressing the ever increasing bioanalytical challenges for biotherapeutics and biomarker assays. The highly specific, selective and sensitive characteristics of the immunocapture-LC-MS-based assays have enabled the determination of biotherapeutics and biomarkers in biomatrices with ease of method development, less requirements on key reagents as well as structural specificity for endogenous and engineered biomolecules. In addition, the versatile immunocapture-LC-MS technology has expanded into many challenging areas to enhance mechanistic studies of drug interactions with their targets. This paper intends to summarize our perspectives on enhancing the use of immunocapture-LC-MS in drug discovery and development for emerging new modalities.
Collapse
|
14
|
Farrokhi V, Chabot JR, Neubert H, Yang Z. Assessing the Feasibility of Neutralizing Osteopontin with Various Therapeutic Antibody Modalities. Sci Rep 2018; 8:7781. [PMID: 29773891 PMCID: PMC5958109 DOI: 10.1038/s41598-018-26187-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 05/08/2018] [Indexed: 12/14/2022] Open
Abstract
Osteopontin is a secreted glycophosphoprotein that is highly implicated in many physiological and pathological processes such as biomineralization, cell-mediated immunity, inflammation, fibrosis, cell survival, tumorigenesis and metastasis. Antibodies against osteopontin have been actively pursued as potential therapeutics for various diseases by pharmaceutical companies and academic laboratories. Many studies have demonstrated the efficacy of osteopontin inhibition in a variety of preclinical models of diseases such as rheumatoid arthritis, cancer, nonalcoholic steatohepatitis, but clinical utility has not yet been demonstrated. To evaluate the feasibility of osteopontin neutralization with antibodies in a clinical setting, we measured its physiological turnover rate in humans, a sensitive parameter required for mechanistic pharmacokinetic and pharmacodynamic (PK/PD) modeling of biotherapeutics. Results from a stable isotope-labelled amino acid pulse-chase study in healthy human subjects followed by mass spectrometry showed that osteopontin undergoes very rapid turnover. PK/PD modeling and simulation of different theoretical scenarios reveal that achieving sufficient target coverage using antibodies can be very challenging mostly due to osteopontin’s fast turnover, as well as its relatively high plasma concentrations in human. Therapeutic antibodies against osteopontin would need to be engineered to have much extended PK than conventional antibodies, and be administered at high doses and with short dosing intervals.
Collapse
Affiliation(s)
- Vahid Farrokhi
- Biomedicine Design, Worldwide Research and Development, Pfizer Inc., Andover, Massachusetts, 01810, USA
| | - Jeffrey R Chabot
- Biomedicine Design, Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts, 02139, USA
| | - Hendrik Neubert
- Biomedicine Design, Worldwide Research and Development, Pfizer Inc., Andover, Massachusetts, 01810, USA
| | - Zhiyong Yang
- Inflammation and Immunology Research Unit, Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts, 02139, USA.
| |
Collapse
|
15
|
Farrokhi V, Chen X, Neubert H. Protein Turnover Measurements in Human Serum by Serial Immunoaffinity LC-MS/MS. Clin Chem 2018; 64:279-288. [DOI: 10.1373/clinchem.2017.272922] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 08/17/2017] [Indexed: 12/15/2022]
Abstract
Abstract
BACKGROUND
The half-life of target proteins is frequently an important parameter in mechanistic pharmacokinetic and pharmacodynamic (PK/PD) modeling of biotherapeutics. Clinical studies for accurate measurement of physiologically relevant protein turnover can reduce the uncertainty in PK/PD model-based predictions, for example, of the therapeutic dose and dosing regimen in first-in-human clinical trials.
METHODS
We used a targeted mass spectrometry work flow based on serial immunoaffinity enrichment ofmultiple human serum proteins from a [5,5,5-2H3]-L-leucine tracer pulse-chase study in healthy volunteers. To confirm the reproducibility of turnover measurements from serial immunoaffinity enrichment, multiple aliquots from the same sample set were subjected to protein turnover analysis in varying order. Tracer incorporation was measured by multiple–reaction-monitoring mass spectrometry and target turnover was calculated using a four-compartment pharmacokinetic model.
RESULTS
Five proteins of clinical or therapeutic relevance including soluble tumor necrosis factor receptor superfamily member 12A, tissue factor pathway inhibitor, soluble interleukin 1 receptor like 1, soluble mucosal addressin cell adhesion molecule 1, and muscle-specific creatine kinase were sequentially subjected to turnover analysis from the same human serum sample. Calculated half-lives ranged from 5–15 h; however, no tracer incorporation was observed for mucosal addressin cell adhesion molecule 1.
CONCLUSIONS
The utility of clinical pulse-chase studies to investigate protein turnover can be extended by serial immunoaffinity enrichment of target proteins. Turnover analysis from serum and subsequently from remaining supernatants provided analytical sensitivity and reproducibility for multiple human target proteins in the same sample set, irrespective of the order of analysis.
Collapse
Affiliation(s)
- Vahid Farrokhi
- Biomedicine Design, Worldwide Research & Development, Pfizer, Inc., Andover, MA
| | - Xiaoying Chen
- Clinical Pharmacology, Worldwide Research & Development, Pfizer, Inc., La Jolla, CA
| | - Hendrik Neubert
- Biomedicine Design, Worldwide Research & Development, Pfizer, Inc., Andover, MA
| |
Collapse
|
16
|
Thomas A, Schänzer W, Thevis M. Immunoaffinity techniques coupled to mass spectrometry for the analysis of human peptide hormones: advances and applications. Expert Rev Proteomics 2017; 14:799-807. [PMID: 28758805 DOI: 10.1080/14789450.2017.1362338] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The accurate and comprehensive determination of peptide hormones from biological fluids has represented a considerable challenge to analytical chemists for decades. Besides long-established bioanalytical ligand binding assays (or ELISA, RIA, etc.), more and more mass spectrometry-based methods have been developed recently for purposes commonly referred to as targeted proteomics. Eventually the combination of both, analyte extraction by immunoaffinity and subsequent detection by mass spectrometry, has shown to synergistically enhance the test methods' performance characteristics. Areas covered: The review provides an overview about the actual state of existing methods and applications concerning the analysis of endogenous peptide hormones. Here, special focus is on recent developments considering the extraction procedures with immobilized antibodies, the subsequent separation of target analytes, and their detection by mass spectrometry. Expert commentary: Key aspects of procedures aiming at the detection and/or quantification of peptidic analytes in biological matrices have experienced considerable improvements in the last decade, particularly in terms of the assays' sensitivity, the option of multiplexing target compounds, automatization, and high throughput operation. Despite these advances and progress as expected to be seen in the near future, immunoaffinity purification coupled to mass spectrometry is not yet a standard procedure in routine analysis compared to ELISA/RIA.
Collapse
Affiliation(s)
- Andreas Thomas
- a Institute of Biochemistry/Center for Preventive Doping Research , German Sport University Cologne , Cologne , Germany
| | - Wilhelm Schänzer
- a Institute of Biochemistry/Center for Preventive Doping Research , German Sport University Cologne , Cologne , Germany
| | - Mario Thevis
- a Institute of Biochemistry/Center for Preventive Doping Research , German Sport University Cologne , Cologne , Germany.,b European Monitoring Center for Emerging Doping Agents (EuMoCEDA) , Cologne/Bonn , Germany
| |
Collapse
|
17
|
Millar JS, Lassman ME, Thomas T, Ramakrishnan R, Jumes P, Dunbar RL, deGoma EM, Baer AL, Karmally W, Donovan DS, Rafeek H, Wagner JA, Holleran S, Obunike J, Liu Y, Aoujil S, Standiford T, Gutstein DE, Ginsberg HN, Rader DJ, Reyes-Soffer G. Effects of CETP inhibition with anacetrapib on metabolism of VLDL-TG and plasma apolipoproteins C-II, C-III, and E. J Lipid Res 2017; 58:1214-1220. [PMID: 28314859 PMCID: PMC5454510 DOI: 10.1194/jlr.m074880] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/16/2017] [Indexed: 01/30/2023] Open
Abstract
Cholesteryl ester transfer protein (CETP) mediates the transfer of HDL cholesteryl esters for triglyceride (TG) in VLDL/LDL. CETP inhibition, with anacetrapib, increases HDL-cholesterol, reduces LDL-cholesterol, and lowers TG levels. This study describes the mechanisms responsible for TG lowering by examining the kinetics of VLDL-TG, apoC-II, apoC-III, and apoE. Mildly hypercholesterolemic subjects were randomized to either placebo (N = 10) or atorvastatin 20 mg/qd (N = 29) for 4 weeks (period 1) followed by 8 weeks of anacetrapib, 100 mg/qd (period 2). Following each period, subjects underwent stable isotope metabolic studies to determine the fractional catabolic rates (FCRs) and production rates (PRs) of VLDL-TG and plasma apoC-II, apoC-III, and apoE. Anacetrapib reduced the VLDL-TG pool on a statin background due to an increased VLDL-TG FCR (29%; P = 0.002). Despite an increased VLDL-TG FCR following anacetrapib monotherapy (41%; P = 0.11), the VLDL-TG pool was unchanged due to an increase in the VLDL-TG PR (39%; P = 0.014). apoC-II, apoC-III, and apoE pool sizes increased following anacetrapib; however, the mechanisms responsible for these changes differed by treatment group. Anacetrapib increased the VLDL-TG FCR by enhancing the lipolytic potential of VLDL, which lowered the VLDL-TG pool on atorvastatin background. There was no change in the VLDL-TG pool in subjects treated with anacetrapib monotherapy due to an accompanying increase in the VLDL-TG PR.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Joseph Obunike
- New York City College of Technology, CUNY, Brooklyn, NY 11201
| | - Yang Liu
- Merck & Co., Inc., Kenilworth, NJ 07033
| | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW Mass spectrometry is an ever evolving technology that is equipped with a variety of tools for protein research. Some lipoprotein studies, especially those pertaining to HDL biology, have been exploiting the versatility of mass spectrometry to understand HDL function through its proteome. Despite the role of mass spectrometry in advancing research as a whole, however, the technology remains obscure to those without hands on experience, but still wishing to understand it. In this review, we walk the reader through the coevolution of common mass spectrometry workflows and HDL research, starting from the basic unbiased mass spectrometry methods used to profile the HDL proteome to the most recent targeted methods that have enabled an unprecedented view of HDL metabolism. RECENT FINDINGS Unbiased global proteomics have demonstrated that the HDL proteome is organized into subgroups across the HDL size fractions providing further evidence that HDL functional heterogeneity is in part governed by its varying protein constituents. Parallel reaction monitoring, a novel targeted mass spectrometry method, was used to monitor the metabolism of HDL apolipoproteins in humans and revealed that apolipoproteins contained within the same HDL size fraction exhibit diverse metabolic properties. SUMMARY Mass spectrometry provides a variety of tools and strategies to facilitate understanding, through its proteins, the complex biology of HDL.
Collapse
Affiliation(s)
- Sasha A. Singh
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Center for Excellence in Vascular Biology, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
19
|
Plasma PCSK9 measurement by liquid chromatography-Tandem mass spectrometry and comparison with conventional ELISA. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1044-1045:24-29. [PMID: 28064066 DOI: 10.1016/j.jchromb.2016.12.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 12/21/2016] [Accepted: 12/30/2016] [Indexed: 11/22/2022]
Abstract
The combination of liquid chromatography-tandem mass spectrometry (LC-MS/MS) and trypsin proteolysis is an effective tool for accurate quantitation of multiple proteins in a single run. However, expensive samples pre-treatment as immunoenrichment are often required to analyze low abundant proteins. Plasma proprotein convertase subtilisin/kexin type 9 (PCSK9), a circulating regulator of low-density lipoprotein metabolism, was studied as an example of a low abundant plasma protein. We investigated post-proteolysis solid-phase extraction (SPE) as an alternative strategy to improve its detection. After optimization of pretreatment, including denaturation, reduction, alkylation, tryptic digestion and selective SPE concentration, 91±7% of PCSK9 was recovered from human plasma samples and coefficients of variation were less than 13.2% with a lower limit of quantification of 37.5ng/ml. This LC-MS/MS method was compared with standard enzyme-linked immunosorbent assay in 30 human plasma samples with a broad range of PCSK9 concentrations. Both methods were significantly correlated (r=0.936, p<0.001) with less than 7% of the values out of the 95% confidence interval and similar concentrations were measured using either LC-MS/MS or ELISA methods (514.2±217.2 vs. 504.2±231.0ng/ml, respectively- p=NS). This method involving SPE is an effective measurement tool for low abundant plasma protein analysis that could be easily included in multiplexed assays.
Collapse
|
20
|
SRM-based measurements of proprotein convertase subtilisin/kexin type 9 and lipoprotein(a) kinetics in nonhuman primate serum. Bioanalysis 2016; 8:2551-2563. [DOI: 10.4155/bio-2016-0146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: PCSK9 and Lp(a) have been identified as potential biomarkers for cardiovascular disease. The ability to measure protein turnover rates will provide insights into the dynamic properties of these proteins and lead to better understanding of their biological roles. We aimed to implement the stable isotope-labeled tracers ([2H3]-leucine) and develop a novel LC-selected reaction monitoring (SRM) mass spectrometry (MS) method to study the kinetics of PCSK9 and Lp(a). Results: A sensitive method using immunoaffinity enrichment coupled with LC-SRM MS was developed to measure the production and degradation rates of PCSK9 and Lp(a) in naive nonhuman primate serum. Comparable results were obtained from two different routes of tracer administration. Conclusion: Immunoaffinity enrichment coupled with LC-SRM MS demonstrated success in in vivo kinetic measurements of proteins with relatively slow turnover rate (Lp[a]) or low abundance (PCSK9) in serum.
Collapse
|
21
|
Abstract
Medical diagnostics and treatment has advanced from a one size fits all science to treatment of the patient as a unique individual. Currently, this is limited solely to genetic analysis. However, epigenetic, transcriptional, proteomic, posttranslational modifications, metabolic, and environmental factors influence a patient’s response to disease and treatment. As more analytical and diagnostic techniques are incorporated into medical practice, the personalized medicine initiative transitions to precision medicine giving a holistic view of the patient’s condition. The high accuracy and sensitivity of mass spectrometric analysis of proteomes is well suited for the incorporation of proteomics into precision medicine. This review begins with an overview of the advance to precision medicine and the current state of the art in technology and instrumentation for mass spectrometry analysis. Thereafter, it focuses on the benefits and potential uses for personalized proteomic analysis in the diagnostic and treatment of individual patients. In conclusion, it calls for a synthesis between basic science and clinical researchers with practicing clinicians to design proteomic studies to generate meaningful and applicable translational medicine. As clinical proteomics is just beginning to come out of its infancy, this overview is provided for the new initiate.
Collapse
|
22
|
The clinical utility of mass spectrometry based protein assays. Clin Chim Acta 2016; 459:155-161. [DOI: 10.1016/j.cca.2016.05.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/25/2016] [Accepted: 05/30/2016] [Indexed: 11/22/2022]
|
23
|
An evaluation of an aptamer for use as an affinity reagent with MS: PCSK9 as an example protein. Bioanalysis 2016; 8:1557-1564. [PMID: 27397798 DOI: 10.4155/bio-2016-0046] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND For quantitative immunoaffinity IA-LC-MS, the utility of antibodies has been demonstrated many times but the utility of aptamers as affinity reagents is unproven. METHODS Immunoaffinity reagents including a monoclonal antibody and an aptamer were coupled to magnetic beads and used as part of an enrichment strategy for PCSK9 quantitation in plasma. RESULTS With limited method development, we have established a comparison of an anti-PCSK9 aptamer with an anti-PCSK9 monoclonal antibody. The background that results from a tryptic digest of affinity enrichment in plasma was demonstrated for each reagent using high-resolution full scan MS. The assay recovery was demonstrated for multiple concentrations of aptamer in plasma with different concentrations of PCSK9 protein. CONCLUSION The aptamer achieved comparable enrichment to the antibody, but with lower peptide background, thus demonstrating the potential use of aptamers for IA-LC-MS.
Collapse
|
24
|
Egeland SV, Reubsaet L, Halvorsen TG. The pros and cons of increased trypsin-to-protein ratio in targeted protein analysis. J Pharm Biomed Anal 2016; 123:155-61. [DOI: 10.1016/j.jpba.2016.02.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 01/29/2016] [Accepted: 02/09/2016] [Indexed: 02/06/2023]
|
25
|
Singh SA, Andraski AB, Pieper B, Goh W, Mendivil CO, Sacks FM, Aikawa M. Multiple apolipoprotein kinetics measured in human HDL by high-resolution/accurate mass parallel reaction monitoring. J Lipid Res 2016; 57:714-28. [PMID: 26862155 DOI: 10.1194/jlr.d061432] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Indexed: 01/10/2023] Open
Abstract
Endogenous labeling with stable isotopes is used to study the metabolism of proteins in vivo. However, traditional detection methods such as GC/MS cannot measure tracer enrichment in multiple proteins simultaneously, and multiple reaction monitoring MS cannot measure precisely the low tracer enrichment in slowly turning-over proteins as in HDL. We exploited the versatility of the high-resolution/accurate mass (HR/AM) quadrupole Orbitrap for proteomic analysis of five HDL sizes. We identified 58 proteins in HDL that were shared among three humans and that were organized into five subproteomes according to HDL size. For seven of these proteins, apoA-I, apoA-II, apoA-IV, apoC-III, apoD, apoE, and apoM, we performed parallel reaction monitoring (PRM) to measure trideuterated leucine tracer enrichment between 0.03 to 1.0% in vivo, as required to study their metabolism. The results were suitable for multicompartmental modeling in all except apoD. These apolipoproteins in each HDL size mainly originated directly from the source compartment, presumably the liver and intestine. Flux of apolipoproteins from smaller to larger HDL or the reverse contributed only slightly to apolipoprotein metabolism. These novel findings on HDL apolipoprotein metabolism demonstrate the analytical breadth and scope of the HR/AM-PRM technology to perform metabolic research.
Collapse
Affiliation(s)
- Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Allison B Andraski
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA
| | - Brett Pieper
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Wilson Goh
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | - Frank M Sacks
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
26
|
Singh SA, Miyosawa K, Aikawa M. Mass spectrometry meets the challenge of understanding the complexity of the lipoproteome: recent findings regarding proteins involved in dyslipidemia and cardiovascular disease. Expert Rev Proteomics 2015; 12:519-32. [PMID: 26325144 DOI: 10.1586/14789450.2015.1078731] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Despite the fact that link between dyslipidemia and atherosclerosis was made over 100 years ago, atherosclerosis remains a major cause of morbidity and mortality worldwide. Major efforts focus towards understanding lipid metabolism, particularly by studying its particle compartments in circulation: the lipoproteins. In recent years, mass spectrometry has played an integral role in the deep sequencing of the lipoproteome and in metabolism studies conducted in vivo. This review highlights the path of lipoprotein research towards state-of-the-art mass spectrometry with special emphasis on the method of selected reaction monitoring and its impact on apolipoprotein metabolism studies. Also presented is what is expected for the lipoprotein field with the recent advent of high resolution/accurate mass parallel reaction monitoring mass spectrometry. The benefits of high resolution/accurate mass measurements are demonstrated by example instrument workflows and by detailing a novel method to quantify very low levels of circulating proprotein convertase subtilisin-kexin type 9 in rabbit. It is anticipated that future clinical studies or clinical trials aimed to treat dyslipidemia by manipulating key regulatory proteins will benefit from the new and exciting opportunities afforded by the latest technologies in mass spectrometry.
Collapse
Affiliation(s)
- Sasha A Singh
- a 1 Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Katsutoshi Miyosawa
- a 1 Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Masanori Aikawa
- a 1 Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,b 2 Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
27
|
Millar JS, Reyes-Soffer G, Jumes P, Dunbar RL, deGoma EM, Baer AL, Karmally W, Donovan DS, Rafeek H, Pollan L, Tohyama J, Johnson-Levonas AO, Wagner JA, Holleran S, Obunike J, Liu Y, Ramakrishnan R, Lassman ME, Gutstein DE, Ginsberg HN, Rader DJ. Anacetrapib lowers LDL by increasing ApoB clearance in mildly hypercholesterolemic subjects. J Clin Invest 2015; 125:2510-22. [PMID: 25961461 DOI: 10.1172/jci80025] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 04/13/2015] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Individuals treated with the cholesteryl ester transfer protein (CETP) inhibitor anacetrapib exhibit a reduction in both LDL cholesterol and apolipoprotein B (ApoB) in response to monotherapy or combination therapy with a statin. It is not clear how anacetrapib exerts these effects; therefore, the goal of this study was to determine the kinetic mechanism responsible for the reduction in LDL and ApoB in response to anacetrapib. METHODS We performed a trial of the effects of anacetrapib on ApoB kinetics. Mildly hypercholesterolemic subjects were randomized to background treatment of either placebo (n = 10) or 20 mg atorvastatin (ATV) (n = 29) for 4 weeks. All subjects then added 100 mg anacetrapib to background treatment for 8 weeks. Following each study period, subjects underwent a metabolic study to determine the LDL-ApoB-100 and proprotein convertase subtilisin/kexin type 9 (PCSK9) production rate (PR) and fractional catabolic rate (FCR). RESULTS Anacetrapib markedly reduced the LDL-ApoB-100 pool size (PS) in both the placebo and ATV groups. These changes in PS resulted from substantial increases in LDL-ApoB-100 FCRs in both groups. Anacetrapib had no effect on LDL-ApoB-100 PRs in either treatment group. Moreover, there were no changes in the PCSK9 PS, FCR, or PR in either group. Anacetrapib treatment was associated with considerable increases in the LDL triglyceride/cholesterol ratio and LDL size by NMR. CONCLUSION These data indicate that anacetrapib, given alone or in combination with a statin, reduces LDL-ApoB-100 levels by increasing the rate of ApoB-100 fractional clearance. TRIAL REGISTRATION ClinicalTrials.gov NCT00990808. FUNDING Merck & Co. Inc., Kenilworth, New Jersey, USA. Additional support for instrumentation was obtained from the National Center for Advancing Translational Sciences (UL1TR000003 and UL1TR000040).
Collapse
|
28
|
Hutcheson JD, Goettsch C, Pham T, Iwashita M, Aikawa M, Singh SA, Aikawa E. Enrichment of calcifying extracellular vesicles using density-based ultracentrifugation protocol. J Extracell Vesicles 2014; 3:25129. [PMID: 25491249 PMCID: PMC4261240 DOI: 10.3402/jev.v3.25129] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 11/07/2014] [Accepted: 11/10/2014] [Indexed: 01/04/2023] Open
Abstract
Calcifying extracellular vesicles (EVs) released from cells within atherosclerotic plaques have received increased attention for their role in mediating vascular calcification, a major predictor of cardiovascular morbidity and mortality. However, little is known about the difference between this pathologic vesicle population and other EVs that contribute to physiological cellular processes. One major challenge that hinders research into these differences is the inability to selectively isolate calcifying EVs from other vesicle populations. In this study, we hypothesized that the formation of mineral within calcifying EVs would increase the density of the vesicles such that they would pellet at a faster rate during ultracentrifugation. We show that after 10 min of ultracentrifugation at 100,000×g, calcifying EVs are depleted from the conditioned media of calcifying coronary artery smooth muscle cells and are enriched in the pelleted portion. We utilized mass spectrometry to establish functional proteomic differences between the calcifying EVs enriched in the 10 min ultracentrifugation compared to other vesicle populations preferentially pelleted by longer ultracentrifugation times. The procedures established in this study will allow us to enrich the vesicle population of interest and perform advanced proteomic analyses to find subtle differences between calcifying EVs and other vesicle populations that may be translated into therapeutic targets for vascular calcification. Finally, we will show that the differences in ultracentrifugation times required to pellet the vesicle populations can also be used to estimate physical differences between the vesicles.
Collapse
Affiliation(s)
- Joshua D Hutcheson
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Claudia Goettsch
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tan Pham
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Masaya Iwashita
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA;
| |
Collapse
|
29
|
Quantitation of human peptides and proteins via MS: review of analytically validated assays. Bioanalysis 2014; 6:1843-57. [DOI: 10.4155/bio.14.145] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Since the development of monoclonal antibodies in the 1970s, antibody-based assays have been used for the quantitation of proteins and peptides and, today, they are the most widely used technology in routine laboratory medicine and bioanalysis. However, in the last couple of decades, liquid chromatography-mass spectrometry/mass spectrometry (LC–MS/MS) techniques have been adopted in the quantitation of small molecules, and more recently have made significant contributions in the quantitation of proteins and peptides. In this article, we will review clinical MS-based assays for endogenous peptides, proteins, and therapeutic antibodies, for which validated methods exist. We will also cover the measurement of protein turnover and the unique solutions that MS can offer in this field.
Collapse
|
30
|
Applications of low-flow LC–SRM for the analysis of large molecules in pharmaceutical R&D. Bioanalysis 2014; 6:1859-67. [DOI: 10.4155/bio.14.141] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Although ligand-binding assays are frequently employed to measure large molecules, the use of LC–SRM assays is increasingly popular due to the inherent selectivity advantage and the ability to operate without exquisitely selective antibodies. Until recently LC–SRM assays have been unable to compete with ligand-binding assays in terms of sensitivity. However, the use of low-flow chromatography prior to mass spectrometry has played a crucial role in increasing the sensitivity of LC–SRM platforms and enabling measurements of large molecules that had previously been unmeasurable. In this article, we highlight some technical advances, describe strategies for employing low-flow chromatography, and review recent literature that describes implementation of low-flow LC–SRM to support large-molecule analysis in pharmaceutical R&D.
Collapse
|